Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
PLoS Negl Trop Dis ; 18(6): e0012216, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38848311

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel tick-borne viral pathogen that causes severe fever with thrombocytopenia syndrome (SFTS). The disease was initially reported in central and eastern China, then later in Japan and South Korea, with a mortality rate of 13-30%. Currently, no vaccines or effective therapeutics are available for SFTS treatment. In this study, three monoclonal antibodies (mAbs) targeting the SFTSV envelope glycoprotein Gn were obtained using the hybridoma technique. Two mAbs recognized linear epitopes and did not neutralize SFTSV, while the mAb 40C10 can effectively neutralized SFTSV of different genotypes and also the SFTSV-related Guertu virus (GTV) and Heartland virus (HRTV) by targeting a spatial epitope of Gn. Additionally, the mAb 40C10 showed therapeutic effect in mice infected with different genotypes of SFTSV strains against death by preventing the development of lesions and by promoting virus clearance in tissues. The therapeutic effect could still be observed in mice infected with SFTSV which were administered with mAb 40C10 after infection even up to 4 days. These findings enhance our understanding of SFTSV immunogenicity and provide valuable information for designing detection methods and strategies targeting SFTSV antigens. The neutralizing mAb 40C10 possesses the potential to be further developed as a therapeutic monoclonal antibody against SFTSV and SFTSV-related viruses.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Camundongos Endogâmicos BALB C , Phlebovirus , Phlebovirus/imunologia , Phlebovirus/genética , Animais , Anticorpos Monoclonais/imunologia , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Feminino , Febre Grave com Síndrome de Trombocitopenia/imunologia , Febre Grave com Síndrome de Trombocitopenia/virologia , Epitopos/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Glicoproteínas/imunologia , Glicoproteínas/genética , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/virologia , Infecções por Bunyaviridae/prevenção & controle , Humanos
2.
Vaccine ; 41(20): 3275-3284, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37085455

RESUMO

Schmallenberg Virus (SBV), an arbovirus from the Peribunyaviridae family and Orthobunyavirus genus, was discovered in late 2011 in Germany and has been circulating in Europe, Asia and Africa ever since. The virus causes a disease associated with ruminants that includes fever, fetal malformation, drop in milk production, diarrhoea and stillbirths, becoming a burden for small and large farms. Building on previous studies on SBV nucleoprotein (SBV-N) as a promising vaccine candidate, we have investigated the possible protein regions responsible for protection. Based on selective truncation of domains designed from the available crystal structure of the SBV-N, we identified both the N-terminal domain (N-term; Met1 - Thr133) and a smaller fragment within (C4; Met1 - Ala58) as vaccine prototypes. Two injections of the N-term and C4 polypeptides protected mice knockout for type I interferon (IFN) receptors (IFNAR-/-) challenged with virulent SBV, opposite to control groups that presented severe signs of morbidity and weight loss. Viremia analyses along with the presence of IFN-γ secreted from splenocytes re-stimulated with the N-terminal region of the protein corroborate that these two portions of SBV-N can be employed as subunit vaccines. Apart from both proteinaceous fragments being easily produced in bacterial cells, the C4 polypeptide shares a high sequence homology (∼87.1 %) with the corresponding region of nucleoproteins of several viruses of the Simbu serogroup, a group of Orthobunyaviruses that comprises SBV and veterinary pathogens like Akabane virus and human infecting viruses like Oropouche. Thus, we propose that this smaller fragment is better suited for vaccine nanoparticle formulation, and it paves the way to further research with other related Orthobunyaviruses.


Assuntos
Infecções por Bunyaviridae , Doenças dos Bovinos , Orthobunyavirus , Vacinas , Humanos , Animais , Camundongos , Bovinos , Orthobunyavirus/genética , Infecções por Bunyaviridae/prevenção & controle , Infecções por Bunyaviridae/veterinária , Viremia/prevenção & controle , Nucleoproteínas/genética , Sorogrupo , Imunização , Ruminantes , Doenças dos Bovinos/prevenção & controle
3.
mBio ; 12(4): e0046321, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34340542

RESUMO

Oropouche virus (OROV) infection of humans is associated with a debilitating febrile illness that can progress to meningitis or encephalitis. First isolated from a forest worker in Trinidad and Tobago in 1955, the arbovirus OROV has since been detected throughout the Amazon basin with an estimated 500,000 human infections over 60 years. Like other members of the family Peribunyaviridae, the viral genome exists as 3 single-stranded negative-sense RNA segments. The medium-sized segment encodes a viral glycoprotein complex (GPC) that is proteolytically processed into two viral envelope proteins, Gn and Gc, responsible for attachment and membrane fusion. There are no therapeutics or vaccines to combat OROV infection, and we have little understanding of protective immunity to infection. Here, we generated a replication competent chimeric vesicular stomatitis virus (VSV), in which the endogenous glycoprotein was replaced by the GPC of OROV. Serum from mice immunized by intramuscular injection with VSV-OROV specifically neutralized wild-type OROV, and using peptide arrays we mapped multiple epitopes within an N-terminal variable region of Gc recognized by the immune sera. VSV-OROV lacking this variable region of Gc was also immunogenic in mice producing neutralizing sera that recognize additional regions of Gc. Challenge of both sets of immunized mice with wild-type OROV shows that the VSV-OROV chimeras reduce wild-type viral infection and suggest that antibodies that recognize the variable N terminus of Gc afford less protection than those that target more conserved regions of Gc. IMPORTANCE Oropouche virus (OROV), an orthobunyavirus found in Central and South America, is an emerging public health challenge that causes debilitating febrile illness. OROV is transmitted by arthropods, and increasing mobilization has the potential to significantly increase the spread of OROV globally. Despite this, no therapeutics or vaccines have been developed to combat infection. Using vesicular stomatitis (VSV) as a backbone, we developed a chimeric virus bearing the OROV glycoproteins (VSV-OROV) and tested its ability to elicit a neutralizing antibody response. Our results demonstrate that VSV-OROV produces a strong neutralizing antibody response that is at least partially targeted to the N-terminal region of Gc. Importantly, vaccination with VSV-OROV reduces viral loads in mice challenged with wild-type virus. These data provide novel evidence that targeting the OROV glycoproteins may be an effective vaccination strategy to combat OROV infection.


Assuntos
Infecções por Bunyaviridae/prevenção & controle , Genoma Viral , Orthobunyavirus/genética , Vesiculovirus/genética , Vesiculovirus/imunologia , Proteínas do Envelope Viral/genética , Animais , Anticorpos Neutralizantes , Infecções por Bunyaviridae/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estomatite Vesicular/virologia , Replicação Viral
4.
J Vet Sci ; 20(6): e58, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31775185

RESUMO

The Schmallenberg virus (SBV) is an orthobunyavirus that causes abortions, stillbirths, and congenital defects in pregnant sheep and cattle. Inactivated or live attenuated vaccines have been developed in endemic countries, but there is still interest in the development of SBV vaccines that would allow Differentiating Infected from Vaccinated Animals (DIVA). Therefore, an attempt was made to develop novel DIVA-compatible SBV vaccines using SBV glycoproteins expressed in baculovirus. All vaccines and phosphate buffered saline (PBS) controls were prepared with adjuvant and administered subcutaneously to cattle at 6 month of age. The first trial included 2 groups of animals vaccinated with either carboxyl-terminus glycoprotein (Gc) or PBS and boosted after 2 weeks. In the second trial, 3 groups of cattle were administered either Gc, Gc and amino-terminus glycoprotein (Gn), or PBS with a booster vaccination after 3 weeks. The animals were challenged with SBV 9 days after the booster vaccination in the first study, and 3 weeks after the booster vaccination in the second study. Using a SBV Gc-specific enzyme-linked immunosorbent assay, antibodies were first detected in serum samples 14 days after the first vaccination in both trials, and peaked on days 7 and 9 after the booster in the first and second trials, respectively. Low titers of neutralizing antibodies were detected in serum from only 3/6 and 2/4 animals in the first and second trial, respectively, at 14 days after the first vaccination. The titers increased 2 to 3-fold after the booster vaccination. SBV-specific RNA was detected in the serum and selective tissues in all animals after SBV challenge independent of vaccination status. The SBV candidate vaccines neither prevented viremia nor conferred protection against SBV infection.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/prevenção & controle , Glicoproteínas/imunologia , Imunogenicidade da Vacina , Orthobunyavirus/fisiologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Infecções por Bunyaviridae/prevenção & controle , Bovinos , Doenças dos Bovinos/imunologia , Distribuição Aleatória , Vacinação/veterinária , Vacinas de Subunidades Antigênicas/imunologia
5.
Vaccine ; 36(34): 5116-5123, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-30049630

RESUMO

Schmallenberg virus (SBV), which emerged in 2011 in Central Europe and subsequently spread very rapidly throughout the continent, affects predominantly ruminants. SBV is transmitted by insect vectors, and therefore vaccination is one of the major tools of disease control. Only recently, a domain connected to virus neutralization has been identified at the amino-terminal part of the viral envelope protein Gc. Here, this Gc domain delivered by recombinant EHV-1 or MVA vector viruses was tested in a vaccination-challenge trial in cattle, one of the major target species of SBV. The EHV-1-based vaccine conferred protection in two of four animals, whereas immunization using the MVA vector vaccine efficiently induced an SBV-specific antibody response and full protection against SBV challenge infection in all the vaccinated animals. Moreover, due to the absence of antibodies against SBVs N-protein, both vector vaccines enable the differentiation between vaccinated and field-infected animals making them to a promising tool to control SBV spread as well as to prevent disease in domestic ruminants.


Assuntos
Infecções por Bunyaviridae/veterinária , Herpesvirus Equídeo 1/genética , Orthobunyavirus/imunologia , Vaccinia virus/genética , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/prevenção & controle , Bovinos , Feminino , Imunogenicidade da Vacina , Vacinação/veterinária , Proteínas do Envelope Viral/genética
6.
Virology ; 511: 175-183, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28865344

RESUMO

Heartland virus (HRTV) is an emerging tick-borne virus (Bunyaviridae, Phlebovirus) that has caused sporadic cases of human disease in several central and mid-eastern states of America. Animal models of HRTV disease are needed to gain insights into viral pathogenesis and advancing antiviral drug development. Presence of clinical disease following HRTV challenge in hamsters deficient in STAT2 function underscores the important role played by type I interferon-induced antiviral responses. However, the recovery of most of the infected animals suggests that other mechanisms to control infection and limit disease offer substantial protection. The most prominent disease sign with HRTV infection in STAT2 knockout hamsters was dramatic weight loss with clinical laboratory and histopathology demonstrating acute inflammation in the spleen, lymph node, liver and lung. Finally, we show that HRTV disease in hamsters can be prevented by the use of favipiravir, a promising broad-spectrum antiviral in clinical development for the treatment of influenza.


Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/prevenção & controle , Pirazinas/uso terapêutico , Fator de Transcrição STAT2/deficiência , Estruturas Animais/patologia , Animais , Quimioprevenção , Cricetinae , Modelos Animais de Doenças , Inflamação/patologia , Interferon Tipo I/imunologia , Resultado do Tratamento
7.
Sci Rep ; 7: 42500, 2017 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-28211908

RESUMO

Schmallenberg virus (SBV) is transmitted by insect vectors, and therefore vaccination is one of the most important tools of disease control. In our study, novel subunit vaccines on the basis of an amino-terminal domain of SBV Gc of 234 amino acids ("Gc Amino") first were tested and selected using a lethal small animal challenge model and then the best performing formulations also were tested in cattle. We could show that neither E. coli expressed nor the reduced form of "Gc Amino" protected from SBV infection. In contrast, both, immunization with "Gc Amino"-encoding DNA plasmids and "Gc-amino" expressed in a mammalian system, conferred protection in up to 66% of the animals. Interestingly, the best performance was achieved with a multivalent antigen containing the covalently linked Gc domains of both, SBV and the related Akabane virus. All vaccinated cattle and mice were fully protected against SBV challenge infection. Furthermore, in the absence of antibodies against the viral N-protein, differentiation between vaccinated and field-infected animals allows an SBV marker vaccination concept. Moreover, the presented vaccine design also could be tested for other members of the Simbu serogroup and might allow the inclusion of additional immunogenic domains.


Assuntos
Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/prevenção & controle , Orthobunyavirus/imunologia , Domínios Proteicos/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Antígenos Virais/imunologia , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/prevenção & controle , Linhagem Celular , Feminino , Imunização , Masculino , Camundongos , Camundongos Knockout , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/química , Vacinas Virais/imunologia
8.
Transbound Emerg Dis ; 64(1): 305-308, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25891033

RESUMO

Schmallenberg virus (SBV) is an emerging Orthobunyavirus affecting European domestic ruminants. In this study, three groups of ewes (n = 3) were inoculated with 1 ml of an SBV infectious serum, via the subcutaneous (SC), intradermal (ID) or intranasal (IN) route. The ewes were monitored for 10 days and no clinical signs were reported. IN inoculation failed to generate any detectable RNAemia. SC and ID inoculation induced typical SBV RNAemia and seroconversion upon day 6 post-inoculation in 3/3 and 2/3 sheep, respectively. In all the animals that showed RNAemia, the viral genome could be detected in spleen and mesenteric lymph nodes. Both the SC and ID routes seem suitable to properly reproduce field conditions, as comparable observations were reported regarding RNAemia, seroconversion and viral genome detection in organs.


Assuntos
Infecções por Bunyaviridae/veterinária , Orthobunyavirus/fisiologia , Doenças dos Ovinos/prevenção & controle , Vacinação/veterinária , Administração Intranasal/veterinária , Animais , Infecções por Bunyaviridae/prevenção & controle , Infecções por Bunyaviridae/virologia , Feminino , Injeções Intradérmicas/veterinária , Injeções Subcutâneas/veterinária , Linfonodos/virologia , Ovinos , Doenças dos Ovinos/virologia , Baço/virologia
9.
Antimicrob Agents Chemother ; 56(8): 4168-74, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22615273

RESUMO

Punta Toro virus (PTV; Bunyaviridae, Phlebovirus) is related to Rift Valley fever virus (RVFV), a pathogenic agent which causes severe disease in humans and livestock primarily in the sub-Saharan region of Africa. The recent range expansion of RVFV and the potential for its intentional release into naïve populations pose a significant threat to public health and agriculture. Studies modeling disease in rodents and nonhuman primates have shown that PTV and RVFV are highly sensitive to the antiviral effects of alpha interferon (IFN-α), an important component of the innate antiviral host response. While recombinant IFN-α has high therapeutic value, its utility for the treatment of neglected tropical diseases is hindered by its short in vivo half-life and costly production of longer-lasting pegylated IFNs. Here, we demonstrate extended preexposure protection against lethal PTV challenge following a single intranasal administration of DEF201, which is a replication-deficient human adenovirus type 5 vector engineered to constitutively express consensus IFN-α (cIFN-α) from transduced host cells. DEF201 was also efficacious when administered within 24 h as a postexposure countermeasure. Serum concentrations of cIFN-α could be detected as early as 8 h following treatment and persisted for more than 1 week. The prolonged antiphlebovirus prophylactic effect, low production costs, and ease of administration make DEF201 a promising agent for intervention during natural disease outbreaks and for countering possible bioterrorist acts.


Assuntos
Adenoviridae/genética , Infecções por Bunyaviridae/prevenção & controle , Interferon-alfa/genética , Interferon-alfa/metabolismo , Phlebovirus , Administração Intranasal , Animais , Antivirais/sangue , Antivirais/metabolismo , Cricetinae , Feminino , Vetores Genéticos , Interferon-alfa/sangue , Fígado/virologia , Mesocricetus , Proteínas Recombinantes
10.
Int Immunopharmacol ; 8(8): 1089-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550012

RESUMO

Recombinant Eimeria antigen (rEA) has been shown to have potent anticancer and antiviral activity in respective mouse disease models, presumably through robust immune stimulation that occurs via TLR11, a pattern recognition receptor that recognizes profilin-like proteins expressed on apicomplexan protozoans. Comparable immunostimulatory activity in other species has yet to be demonstrated. Since rEA is known to be highly effective in treating Punta Toro virus (PTV) infection in mice, its ability to elicit protective immunity in the hamster PTV infection model was investigated. rEA was given alone, or in combination with IL-18 or IL-2, and virally challenged hamsters were observed for mortality. Cytokine transcript profiles for IL-12p40, IL-21, IFN-gamma and TNF-alpha were assessed to evaluate the induction of these inflammatory mediators known to be induced in mice following exposure to rEA. A dose of 100 microg of rEA, given once 4 h prior to viral challenge, and a second time on day 3 of the infection, was found to be the most effective prophylactic therapy protecting 60% of treated hamsters from mortality, compared to only 5-10% observed in animals receiving placebo. Increased expression of IFN-gamma and IL-12p40 was evident following treatment with rEA. The data suggest that rEA does induce host antiviral responses in hamsters that result in significant protection from death, although determining the most appropriate dose for intervention in other species, including humans, will likely be challenging.


Assuntos
Antígenos de Protozoários/imunologia , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/prevenção & controle , Eimeria/imunologia , Phlebovirus/imunologia , Animais , Antígenos de Protozoários/administração & dosagem , Infecções por Bunyaviridae/mortalidade , Cricetinae , Feminino , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Subunidade p40 da Interleucina-12/genética , Interleucina-18/administração & dosagem , Interleucina-18/imunologia , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Mesocricetus , Proibitinas , Proteínas Recombinantes/imunologia , Transcrição Gênica
11.
Antimicrob Agents Chemother ; 50(6): 2023-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723561

RESUMO

A protein antigen from an Eimeria protozoan has recently been reported to induce antitumor activity in mice. This activity most likely results from the strong induction of interkeukin-12 (IL-12) and gamma interferon (IFN-gamma), which are also essential factors in the establishment of protective immunity against viral infection. We evaluated recombinant Eimeria antigen (rEA) as a potential immunotherapeutic agent in mouse and hamster models of acute phleboviral disease. Punta Toro virus (PTV) was highly sensitive to a single dose of nanogram quantities of rEA in the mouse infection model. Intraperitoneal treatment with rEA also reduced virus load and liver damage associated with PTV infection. IL-12 was elicited following exposure of uninfected mice to quantities of rEA of 10 ng or greater, and the levels peaked at between 3 and 8 h postexposure. IFN-gamma release was induced more slowly and required less rEA (1 ng) to produce a significant rise in systemic levels. The induction of IL-12 and IFN-gamma involved in the coordination of innate and adaptive immune responses to microbial pathogens required myeloid differentiation factor 88, a signaling adaptor shared by most members of the Toll-like receptor (TLR) family. Despite encouraging results in the murine system, rEA failed to protect hamsters challenged with PTV. Our findings suggest that hamsters may lack functional TLR11, which has recently been shown to recognize a profilin-like protein homologous to rEA from the protozoan Toxoplasma gondii. Further investigation into the immunostimulatory capacity of rEA in other mammalian systems is necessary.


Assuntos
Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/prevenção & controle , Eimeria/imunologia , Phlebovirus/imunologia , Proteínas de Protozoários/imunologia , Doença Aguda , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Infecções por Bunyaviridae/mortalidade , Infecções por Bunyaviridae/virologia , Células Cultivadas , Cricetinae , Relação Dose-Resposta Imunológica , Eimeria/genética , Eimeria/crescimento & desenvolvimento , Feminino , Interferon gama/biossíntese , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/biossíntese , Interleucina-12/genética , Interleucina-12/imunologia , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide , Phlebovirus/genética , Phlebovirus/patogenicidade , Proteínas de Protozoários/genética , Proteínas Recombinantes/imunologia , Análise de Sobrevida , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Carga Viral
12.
Antiviral Res ; 69(3): 165-72, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16430975

RESUMO

Cationic liposome-DNA complexes (CLDC) have been demonstrated to induce potent antitumor activities. The ability of these complexes to elicit protective immunity against viral infections has not been fully explored. Here we report findings on the use of CLDC as an antiviral agent in a mouse model of acute phleboviral (Punta Toro virus) disease. CLDC treatment of mice challenged with Punta Toro virus (PTV) resulted in dramatic increases in survival and reduced viral burden and other parameters indicative of protection against disease. CLDC were effective when administered by intraperitoneal and intravenous routes and elicited protective immunity when given within 1 day of virus challenge. Treatments administered 36 h or longer after challenge, however, were not effective in preventing mortality or disease. CLDC treatment induced release of a number of potential antiviral cytokines including IFN-gamma, IL-12, and IFN-alpha. Taken together, our findings indicate that non-specific immunotherapy with CLDC appears to be an effective treatment for blocking PTV-induced disease and suggests that further exploration in other viral disease models may be warranted.


Assuntos
Adjuvantes Imunológicos , Infecções por Bunyaviridae/prevenção & controle , Infecções por Bunyaviridae/terapia , DNA Viral/imunologia , Lipossomos/imunologia , Phlebovirus/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/virologia , Citocinas/sangue , DNA Viral/administração & dosagem , Feminino , Injeções Intraperitoneais , Injeções Intravenosas , Lipossomos/administração & dosagem , Fígado/virologia , Camundongos , Análise de Sobrevida , Carga Viral
13.
In. Travassos da Rosa, Amelia P. A; Vasconcelos, Pedro F. C; Travassos da Rosa, Jorge F. S. An Overview of Arbovirology in Brazil and Neighbouring Countries. Belem, Instituto Evandro Chagas, 1998. p.186-92, tab.
Monografia em Inglês | LILACS | ID: lil-248907
14.
Virology ; 167(2): 442-50, 1988 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2462308

RESUMO

Hybridomas producing monoclonal antibodies to the three major structural proteins of Punta Toro virus (PTV) were established by fusion of spleen cells with Sp2/0-Ag-14 mouse plasmacytoma cells. Thirty-six independently derived monoclonal antibodies were evaluated in neutralization, hemagglutination inhibition, and ELISA assays and the isotype, antigen specificities, and cross-reactivities were determined. These antibodies were also assessed for their ability to provide protection in a murine model. Both G1- and G2-specific antibodies were obtained which neutralized virus infectivity in vitro and inhibited hemagglutination, whereas nucleocapsid-specific antibodies exhibited neither activity. All of the anti-G1 antibodies were PTV-specific, whereas anti-G2 and anti-nucleocapsid antibodies exhibited varying patterns of cross-reactivity with heterologous phleboviruses. All of the G1-reactive monoclonal antibodies, which bound to epitopes in two distinct topological sites as determined by competitive binding assays, provided efficient protection to both immunocompetent and immunosuppressed mice. In contrast, of the 23 G2-reactive antibodies, only 8 were able to protect immunocompetent mice and only one was able to protect immunosuppressed animals. The degree of protection achieved in vivo did not correlate directly with the neutralization titers determined in vitro.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Infecções por Bunyaviridae/prevenção & controle , Bunyaviridae/imunologia , Proteínas Virais/imunologia , Especificidade de Anticorpos , Capsídeo/imunologia , Reações Cruzadas , Epitopos , Glicoproteínas/imunologia , Testes de Inibição da Hemaglutinação , Hibridomas , Imunização Passiva , Isotipos de Imunoglobulinas/análise , Testes de Neutralização , Proteínas do Core Viral/imunologia , Proteínas do Envelope Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA